Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Brain Behav Immun Health ; 32: 100663, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37503358

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease. Most cases of AD are considered idiopathic and likely due to a combination of genetic, environmental, and lifestyle-related risk factors. Despite occurring decades before the typical age of an AD diagnosis, early-life stress (ELS) has been suggested to have long-lasting effects that may contribute to AD risk and pathogenesis. Still, the mechanisms that underlie the role of ELS on AD risk remain largely unknown. Here, we used 5xFAD transgenic mice to study relatively short-term alterations related to ELS in an AD-like susceptible mouse model at 6 weeks of age. To model ELS, we separated pups from their dams for 3 h per day from postnatal day 2-14. Around 6 weeks of age, we found that maternally separated (MS) 5xFAD mice, particularly female mice, displayed increased amyloid-ß-immunoreactivity in the anterior cingulate cortex (ACC) and basolateral amygdala (BLA). In anterior cingulate cortex, we also noted significantly increased intraneuronal amyloid-ß-immunoreactivity associated with MS but only in female mice. Moreover, IBA1-positive DAPI density was significantly increased in relation to MS in ACC and BLA, and microglia in BLA of MS mice had significantly different morphology compared to microglia in non-MS 5xFAD mice. Cytokine analysis showed that male MS mice, specifically, had increased levels of neuroinflammatory markers CXCL1 and IL-10 in hippocampal extracts compared to non-MS counterparts. Additionally, hippocampal extracts from both male and female MS 5xFAD mice had decreased levels of synapse- and activity-related markers Bdnf, 5htr6, Cox2, and Syp in hippocampus. Lastly, we performed behavioral tests to evaluate anxiety- and depressive-like behavior and working memory but could not detect any significant differences between groups. Overall, we detected several sex-specific molecular and cellular alterations in 6-week-old adolescent 5xFAD mice associated with MS that may help explain the connection between ELS and AD risk.

2.
J Neuroinflammation ; 19(1): 151, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35705972

ABSTRACT

BACKGROUND: The risk of developing Alzheimer's disease (AD) is modulated by genetic and environmental factors. Early-life stress (ELS) exposure during critical periods of brain development can impact later brain function and health, including increasing the risk of developing AD. Microglial dysfunction and neuroinflammation have been implicated as playing a role in AD pathology and may be modulated by ELS. To complicate matters further, sex-specific effects have been noted in response to ELS and in the incidence and progression of AD. METHODS: Here, we subjected male and female mice with either a wild type or 5xFAD familial AD-model background to maternal separation (MS) from postnatal day 2 to 14 to induce ELS. RESULTS: We detected hippocampal neuroinflammatory alterations already at postnatal day 15. By 4 months of age, MS mice presented increased immobility time in the forced swim test and a lower discrimination index in the novel object recognition memory test compared to controls. We found altered Bdnf and Arc expression in the hippocampus and increased microglial activation in the prefrontal cortex due to MS in a sex-dependent manner. In 5xFAD mice specifically, MS exacerbated amyloid-beta deposition, particularly in females. In the periphery, the immune cell population was altered by MS exposure. CONCLUSION: Overall, our results demonstrate that MS has both short- and long-term effects on brain regions related to memory and on the inflammatory system, both in the brain and periphery. These ELS-related effects that are detectable even in adulthood may exacerbate pathology and increase the risk of developing AD via sex-specific mechanisms.


Subject(s)
Alzheimer Disease , Brain , Sex Factors , Stress, Psychological , Animals , Female , Male , Mice , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/immunology , Brain/metabolism , Disease Models, Animal , Maternal Deprivation , Mice, Transgenic
3.
Nat Commun ; 8: 14726, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287086

ABSTRACT

Reducing levels of the aggregation-prone Aß peptide that accumulates in the brain with Alzheimer's disease (AD) has been a major target of experimental therapies. An alternative approach may be to stabilize the physiological conformation of Aß. To date, the physiological state of Aß in brain remains unclear, since the available methods used to process brain tissue for determination of Aß aggregate conformation can in themselves alter the structure and/or composition of the aggregates. Here, using synchrotron-based Fourier transform infrared micro-spectroscopy, non-denaturing gel electrophoresis and conformational specific antibodies we show that the physiological conformations of Aß and amyloid precursor protein (APP) in brain of transgenic mouse models of AD are altered before formation of amyloid plaques. Furthermore, focal Aß aggregates in brain that precede amyloid plaque formation localize to synaptic terminals. These changes in the states of Aß and APP that occur prior to plaque formation may provide novel targets for AD therapy.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/ultrastructure , Amyloid beta-Protein Precursor/ultrastructure , Brain/diagnostic imaging , Peptide Fragments/ultrastructure , Plaque, Amyloid/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression , Humans , Mice , Mice, Transgenic , Native Polyacrylamide Gel Electrophoresis , Neurons/metabolism , Neurons/pathology , Neuropeptides/genetics , Neuropeptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presynaptic Terminals , Primary Cell Culture , Protein Aggregates , Protein Conformation , Spectroscopy, Fourier Transform Infrared , Synaptophysin/genetics , Synaptophysin/metabolism , Synchrotrons
4.
Histol Histopathol ; 17(1): 239-46, 2002 01.
Article in English | MEDLINE | ID: mdl-11813874

ABSTRACT

Alzheimer's disease (AD) neuropathology is characterized by accumulation of "senile" plaques (SPs) and neurofibrillary tangles (NFTs) in vulnerable brain regions. SPs are principally composed of aggregates of up to 42/43 amino acid beta-amyloid (A beta) peptides. The discovery of familial AD (FAD) mutations in the genes for the amyloid precursor protein (APP) and presenilins (PSs), all of which increase A beta42 production, support the view that A beta is centrally involved in the pathogenesis of AD. A beta42 aggregates readily, and is thought to seed the formation of fibrils, which then act as templates for plaque formation. A beta is generated by the sequential intracellular cleavage of APP by beta-secretase to generate the N-terminal end of A beta, and intramembranous cleavage by gamma-secretase to generate the C-terminal end. Cell biological studies have demonstrated that A beta is generated in the ER, Golgi, and endosomal/lysosomal system. A central question involving the role of A beta in AD concerns how A beta causes disease and whether it is extracellular A beta deposition and/or intracellular A beta accumulation that initiates the disease process. The most prevalent view is that SPs are composed of extracellular deposits of secreted A beta and that A beta causes toxicity to surrounding neurons as extracellular SP. The recent emphasis on the intracellular biology of APP and A beta has led some investigators to consider the possibility that intraneuronal A beta may directly cause toxicity. In this review we will outline current knowledge of the localization of both intracellular and extracellular A beta.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/metabolism , Humans , Plaque, Amyloid/pathology
5.
Neuron ; 30(3): 641-2, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11430794

ABSTRACT

Transgenic mice developing beta-amyloid (Abeta) plaques are advancing experimental treatment strategies for Alzheimer's disease. The metal chelator, clioquinol, is reported by Cherny et al. (2001) to reduce Abeta plaques, presumably by chelation of Abeta-associated zinc and copper. This and other recent Abeta-modulating treatment approaches are discussed.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Chelating Agents/pharmacology , Clioquinol/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
6.
J Neurosci ; 21(8): 2561-70, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11306609

ABSTRACT

Alzheimer's Disease (AD) is characterized by cerebral accumulation of beta-amyloid peptides (Abeta), which are proteolytically derived from beta-amyloid precursor protein (betaAPP). betaAPP metabolism is highly regulated via various signal transduction systems, e.g., several serine/threonine kinases and phosphatases. Several growth factors known to act via receptor tyrosine kinases also have been demonstrated to regulate sbetaAPP secretion. Among these receptors, insulin and insulin-like growth factor-1 receptors are highly expressed in brain, especially in hippocampus and cortex. Emerging evidence indicates that insulin has important functions in brain regions involved in learning and memory. Here we present evidence that insulin significantly reduces intracellular accumulation of Abeta and that it does so by accelerating betaAPP/Abeta trafficking from the trans-Golgi network, a major cellular site for Abeta generation, to the plasma membrane. Furthermore, insulin increases the extracellular level of Abeta both by promoting its secretion and by inhibiting its degradation via insulin-degrading enzyme. The action of insulin on betaAPP metabolism is mediated via a receptor tyrosine kinase/mitogen-activated protein (MAP) kinase kinase pathway. The results suggest cell biological and signal transduction mechanisms by which insulin modulates betaAPP and Abeta trafficking in neuronal cultures.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Insulin/metabolism , MAP Kinase Signaling System/physiology , Neurons/metabolism , Alzheimer Disease/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Space/metabolism , Golgi Apparatus/metabolism , Insulin/pharmacology , Insulysin , MAP Kinase Signaling System/drug effects , Neurons/cytology , Neurons/drug effects , Peptide Fragments/metabolism , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism
7.
8.
Front Biosci ; 5: D72-83, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10702374

ABSTRACT

In molecular neurobiology, perhaps no molecule has been as thoroughly examined as Alzheimer's beta-amyloid precursor protein (beta-APP). In the years since the cDNA encoding beta-APP was cloned, the protein has been the subject of unparalleled scrutiny on all levels. From molecular genetics and cellular biology to neuroanatomy and epidemiology, no scientific discipline has been left unexplored - and with good reason. beta-amyloid (Abeta) is the main constituent of the amyloidogenic plaques which are a primary pathological hallmark of Alzheimer's disease, and bta-APP is the protein from which Abeta is cleaved and released. Unraveling the molecular events underlying Abeta generation has been, and remains, of paramount importance to scientists in our field. In this review we will trace the progress that has been made in understanding the molecular and cellular basis of beta-APP trafficking and processing, or alternatively stated, the molecular basis for Abeta generation. Imperative to a complete understanding of Abeta generation is the delineation of its subcellular localization and the identification of proteins that play either direct or accessory roles in Abeta generation. We will focus on the regulation of beta-APP cleavage through diverse signal transduction mechanisms and discuss possible points of therapeutic intercession in what has been postulated to be a seminal molecular step in the cascade of events terminating in the onset of dementia, loss of neurons, and eventual death from Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Endopeptidases , Humans , Membrane Proteins/metabolism , Presenilin-2
9.
Proc Natl Acad Sci U S A ; 97(3): 1202-5, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10655508

ABSTRACT

Alzheimer's disease (AD) is characterized by the age-related deposition of beta-amyloid (Abeta) 40/42 peptide aggregates in vulnerable brain regions. Multiple levels of evidence implicate a central role for Abeta in the pathophysiology of AD. Abeta peptides are generated by the regulated cleavage of an approximately 700-aa Abeta precursor protein (betaAPP). Full-length betaAPP can undergo proteolytic cleavage either within the Abeta domain to generate secreted sbetaAPPalpha or at the N- and C-terminal domain(s) of Abeta to generate amyloidogenic Abeta peptides. Several epidemiological studies have reported that estrogen replacement therapy protects against the development of AD in postmenopausal women. We previously reported that treating cultured neurons with 17beta-estradiol reduced the secretion of Abeta40/42 peptides, suggesting that estrogen replacement therapy may protect women against the development of AD by regulating betaAPP metabolism. Increasing evidence indicates that testosterone, especially bioavailable testosterone, decreases with age in older men and in postmenopausal women. We report here that treatment with testosterone increases the secretion of the nonamyloidogenic APP fragment, sbetaAPPalpha, and decreases the secretion of Abeta peptides from N2a cells and rat primary cerebrocortical neurons. These results raise the possibility that testosterone supplementation in elderly men may be protective in the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Testosterone/pharmacology , Alzheimer Disease/prevention & control , Amino Acid Sequence , Animals , Cholesterol/pharmacology , Corticosterone/pharmacology , Estradiol/pharmacology , Female , Humans , Male , Mice , Molecular Sequence Data , Neurons/metabolism , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Secretory Rate/drug effects
10.
Am J Pathol ; 156(1): 15-20, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623648

ABSTRACT

Alzheimer's disease (AD) is characterized by the deposition of senile plaques (SPs) and neurofibrillary tangles (NFTs) in vulnerable brain regions. SPs are composed of aggregated beta-amyloid (Abeta) 40/42(43) peptides. Evidence implicates a central role for Abeta in the pathophysiology of AD. Mutations in betaAPP and presenilin 1 (PS1) lead to elevated secretion of Abeta, especially the more amyloidogenic Abeta42. Immunohistochemical studies have also emphasized the importance of Abeta42 in initiating plaque pathology. Cell biological studies have demonstrated that Abeta is generated intracellularly. Recently, endogenous Abeta42 staining was demonstrated within cultured neurons by confocal immunofluorescence microscopy and within neurons of PS1 mutant transgenic mice. A central question about the role of Abeta in disease concerns whether extracellular Abeta deposition or intracellular Abeta accumulation initiates the disease process. Here we report that human neurons in AD-vulnerable brain regions specifically accumulate gamma-cleaved Abeta42 and suggest that this intraneuronal Abeta42 immunoreactivity appears to precede both NFT and Abeta plaque deposition. This study suggests that intracellular Abeta42 accumulation is an early event in neuronal dysfunction and that preventing intraneuronal Abeta42 aggregation may be an important therapeutic direction for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Adult , Aged , Aged, 80 and over , Brain/pathology , Cadaver , Cognition Disorders/metabolism , Cognition Disorders/pathology , Dementia/metabolism , Dementia/pathology , Dementia/psychology , Down Syndrome/metabolism , Down Syndrome/pathology , Humans , Immunohistochemistry , Infant , Middle Aged , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Reference Values
11.
Proc Natl Acad Sci U S A ; 96(2): 742-7, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892704

ABSTRACT

The excessive generation and accumulation of 40- and 42-aa beta-amyloid peptides (Abeta40/Abeta42) in selectively vulnerable brain regions is a major neuropathological feature of Alzheimer's disease. Abeta, derived by proteolytic cleavage from the beta-amyloid precursor protein (betaAPP), is normally secreted. However, recent evidence suggests that significant levels of Abeta also may remain inside cells. Here, we have investigated the subcellular compartments within which distinct amyloid species are generated and the compartments from which they are secreted. Three experimental approaches were used: (i) immunofluorescence performed in intact cortical neurons; (ii) sucrose gradient fractionation performed with mouse neuroblastoma cells stably expressing wild-type betaAPP695 (N2a695); and (iii) cell-free reconstitution of Abeta generation and trafficking from N2a695 cells. These studies demonstrate that: (i) Abeta40 (Abeta1-40 plus Abetax-40, where x is an NH2-terminal truncation) is generated exclusively within the trans-Golgi Network (TGN) and packaged into post-TGN secretory vesicles; (ii) Abetax-42 is made and retained within the endoplasmic reticulum in an insoluble state; (iii) Abeta42 (Abeta1-42 plus Abetax-42) is made in the TGN and packaged into secretory vesicles; and (iv) the amyloid peptides formed in the TGN consist of two pools (a soluble population extractable with detergents and a detergent-insoluble form). The identification of the organelles in which distinct forms of Abeta are generated and from which they are secreted should facilitate the identification of the proteolytic enzymes responsible for their formation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Animals , Brain/metabolism , Cell Fractionation , Cells, Cultured , Centrifugation, Density Gradient , Humans , Immunohistochemistry , Microscopy, Fluorescence , Peptide Fragments/metabolism , Rats
12.
J Neurochem ; 71(5): 1920-5, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9798916

ABSTRACT

Studies of processing of the Alzheimer beta-amyloid precursor protein (betaAPP) have been performed to date mostly in continuous cell lines and indicate the existence of two principal metabolic pathways: the "beta-secretase" pathway, which generates beta-amyloid (A beta(1-40/42); approximately 4 kDa), and the "alpha-secretase" pathway, which generates a smaller fragment, the "p3" peptide (A beta(17-40/42); approximately 3 kDa). To determine whether similar processing events underlie betaAPP metabolism in neurons, media were examined following conditioning by primary neuronal cultures derived from embryonic day 17 rats. Immunoprecipitates of conditioned media derived from [35S]methionine pulse-labeled primary neuronal cultures contained 4- and 3-kDa A beta-related species. Radiosequencing analysis revealed that the 4-kDa band corresponded to conventional A beta beginning at position A beta(Asp1), whereas both radiosequencing and immunoprecipitation-mass spectrometry analyses indicated that the 3-kDa species in these conditioned media began with A beta(Glu11) at the N terminus, rather than A beta(Leu17) as does the conventional p3 peptide. Either activation of protein kinase C or inhibition of protein phosphatase 1/2A increased soluble betaAPP(alpha) release and decreased generation of both the 4-kDa A beta and the 3-kDa N-truncated A beta. Unlike results obtained with continuously cultured cells, protein phosphatase 1/2A inhibitors were more potent at reducing A beta secretion by neurons than were protein kinase C activators. These data indicate that rodent neurons generate abundant A beta variant peptides and emphasize the role of protein phosphatases in modulating neuronal A beta generation.


Subject(s)
Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Genetic Variation/physiology , Neurons/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Mice , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
13.
Nat Med ; 4(4): 447-51, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9546791

ABSTRACT

Alzheimer's disease (AD) is characterized by the accumulation of cerebral plaques composed of 40- and 42-amino acid beta-amyloid (Abeta) peptides, and autosomal dominant forms of AD appear to cause disease by promoting brain Abeta accumulation. Recent studies indicate that postmenopausal estrogen replacement therapy may prevent or delay the onset of AD. Here we present evidence that physiological levels of 17beta-estradiol reduce the generation of Abeta by neuroblastoma cells and by primary cultures of rat, mouse and human embryonic cerebrocortical neurons. These results suggest a mechanism by which estrogen replacement therapy can delay or prevent AD.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Cerebral Cortex/cytology , Estradiol/pharmacology , Neurons/physiology , Alzheimer Disease , Animals , Cells, Cultured , Coculture Techniques , Embryo, Mammalian , Fetus , Humans , Mice , Neuroblastoma , Neurons/cytology , Neurons/drug effects , Peptide Fragments/biosynthesis , Rats , Recombinant Proteins/biosynthesis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection , Tumor Cells, Cultured
14.
Front Biosci ; 3: d399-407, 1998 Mar 26.
Article in English | MEDLINE | ID: mdl-9514984

ABSTRACT

In molecular neurobiology, perhaps no molecule has been as thoroughly examined as Alzheimer's beta-amyloid precursor protein (betaAPP). In the ten years since the cDNA encoding betaAPP was cloned, the protein has been the subject of unparalleled scrutiny on all levels. From molecular genetics and cellular biology to neuroanatomy and epidemiology, no scientific discipline has been left unexplored - and with good reason. beta-amyloid (Abeta) is the main constituent of the amyloidogenic plaques which are a primary pathological hallmark of Alzheimer's disease, and betaAPP is the protein from which Abeta is cleaved and released. Unraveling the molecular events underlying Abeta generation has been, and remains, of paramount importance to scientists in our field. In this review we will trace the progress that has been made in understanding the molecular and cellular basis of betaAPP trafficking and processing, or alternatively stated, the molecular basis for Abeta generation. Imperative to a complete understanding of Abeta generation is the delineation of its subcellular localization and the identification of proteins which play either direct or accessory roles in Abeta generation. We will focus on the regulation of betaAPP cleavage through diverse signal transduction mechanisms and discuss possible points of therapeutic intercession in what has been postulated to be a seminal molecular step in the cascade of events terminating in the onset of dementia, a loss of neurons, and tragically, eventual death from Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain Chemistry/physiology , Animals , Humans , Neurons/pathology
15.
Proc Natl Acad Sci U S A ; 94(10): 5090-4, 1997 May 13.
Article in English | MEDLINE | ID: mdl-9144195

ABSTRACT

Pathogenic mutations in presenilin 1 (PS1) are associated with approximately 50% of early-onset familial Alzheimer disease. PS1 is endoproteolytically cleaved to yield a 30-kDa N-terminal fragment (NTF) and an 18-kDa C-terminal fragment (CTF). Using COS7 cells transfected with human PS1, we have found that phorbol 12, 13-dibutyrate and forskolin increase the state of phosphorylation of serine residues of the human CTF. Phosphorylation of the human CTF resulted in a shift in electrophoretic mobility from a single major species of 18 kDa to a doublet of 20-23 kDa. This mobility shift was also observed with human PS1 that had been transfected into mouse neuroblastoma (N2a) cells. Treatment of the phosphorylated CTF doublet with phage lambda protein phosphatase eliminated the 20- to 23-kDa doublet while enhancing the 18-kDa species, consistent with the interpretation that the electrophoretic mobility shift was due to the addition of phosphate to the 18-kDa species. The NTF and CTF eluted from a gel filtration column at an estimated mass of over 100 kDa, suggesting that these fragments exist as an oligomerized species. Upon phosphorylation of the PS1 CTF, the apparent mass of the NTF- or CTF-containing oligomers was unchanged. Thus, the association of PS1 fragments may be maintained during cycles of phosphorylation/dephosphorylation of the PS1 CTF.


Subject(s)
Membrane Proteins/biosynthesis , Animals , COS Cells , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Humans , Macromolecular Substances , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Mice , Neuroblastoma , PC12 Cells , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/biosynthesis , Phosphoproteins/chemistry , Phosphoproteins/isolation & purification , Phosphorylation , Presenilin-1 , Protein Kinase Inhibitors , Protein Kinases/metabolism , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Serine , Transfection , Tumor Cells, Cultured
16.
Ann Neurol ; 41(3): 402-4, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9066363

ABSTRACT

Inheritance of the apolipoprotein E (ApoE) epsilon 4 allele is a risk factor for Alzheimer's disease (AD) and is associated with increased deposition of beta-amyloid (A beta) in AD, Down's syndrome, and normal aging. A beta deposition in the form of senile plaques (SPs) has recently been described in patients with temporal lobe epilepsy (TLE). We studied the relationship between ApoE epsilon 4 genotype and the deposition of A beta in temporal lobe tissue from patients who underwent temporal lobectomy for intractable epilepsy. TLE patients with SPs had a 70% ApoE epsilon 4 carrier frequency compared with a 27% carrier frequency among age-matched TLE controls without SPs. Our data suggest that the association between ApoE epsilon 4 and intracerebral A beta accumulation is not unique to the elderly or to those with dementia, and may be a feature of conditions in which there is both an ApoE epsilon 4 allele and over-production of A beta precursor protein, and, presumably, A beta.


Subject(s)
Apolipoproteins E/genetics , Epilepsy, Temporal Lobe/genetics , Amyloid beta-Peptides/analysis , Apolipoprotein E4 , Biopsy , Case-Control Studies , Chi-Square Distribution , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/surgery , Genotype , Humans , Middle Aged , Temporal Lobe/chemistry , Temporal Lobe/pathology
18.
Neurol Res ; 18(4): 291-4; discussion 295-6, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8875444

ABSTRACT

Post-mortem studies of the brain of an Alzheimer patient indicate fewer senile plaques in the crests of cortical gyri underneath an omental transposition than in neighboring cortical areas.


Subject(s)
Alzheimer Disease/pathology , Occipital Lobe/pathology , Omentum/transplantation , Parietal Lobe/pathology , Transplantation, Heterotopic , Alzheimer Disease/surgery , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Atrophy , Cell Hypoxia , Cerebrovascular Circulation , Hemosiderin/analysis , Humans , Iron/metabolism , Male , Occipital Lobe/blood supply , Parietal Lobe/blood supply
19.
Muscle Nerve ; 19(7): 870-5, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8965841

ABSTRACT

Colchicine may induce a myoneuropathy in patients with renal insufficiency. To date, myotonia has not been described in this disorder. We recently studied 4 patients treated with routine doses of colchicine who, in the setting of renal insufficiency, developed a severe myoneuropathy characterized by prominent myotonic discharges on electromyography. In addition, 1 of the 4 patients had profound clinical myotonia. In the 3 patients in whom biopsies were performed, marked myopathic change with intracytoplasmic vacuolization was identified. All 4 patients improved rapidly with discontinuation of the medication. The patient in whom electrophysiologic studies were repeated had a complete resolution of the myotonic discharges. Colchicine myoneuropathy can present with prominent clinical and electrophysiologic myotonia that resolves completely with discontinuation of the medication.


Subject(s)
Colchicine/adverse effects , Myotonia/chemically induced , Neuromuscular Diseases/chemically induced , Adult , Aged , Aged, 80 and over , Colchicine/therapeutic use , Electromyography , Female , Humans , Kidney Failure, Chronic/drug therapy , Male , Middle Aged , Muscles/pathology , Myotonia/pathology , Myotonia/physiopathology , Neuromuscular Diseases/pathology , Neuromuscular Diseases/physiopathology
20.
Arch Ophthalmol ; 114(4): 429-32, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8602780

ABSTRACT

OBJECTIVE: To describe two patients with mesencephalic midline clefts and associated eye movement disorders. DESIGN: Case reports. RESULTS: The first patient developed bilateral internuclear ophthalmoplegia with exotropia, reduced convergence, right ptosis, right fourth-nerve palsy, and right elevator palsy several years after meningitis with hydrocephalus. The second patient had bilateral internuclear ophthalmoplegia with exotropia, reduced convergence, bilateral ptosis, limited upward gaze, and right hypertropia since childhood. In both patients, magnetic resonance imaging showed a midline cleft extending from the cerebral aqueduct into the midbrain. CONCLUSION: It is likely that the clefts affected the oculomotor nuclei and medial longitudinal fasciculi, accounting for the eye movement disorders.


Subject(s)
Brain Diseases/complications , Eye Movements , Mesencephalon/pathology , Ocular Motility Disorders/etiology , Adult , Blepharoptosis/etiology , Brain Diseases/diagnosis , Convergence, Ocular , Exotropia/etiology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Ophthalmoplegia/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...